Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Sci Rep ; 9(1): 19613, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31873100

ABSTRACT

Plants adapted to extreme conditions can be at high risk from climate change; arctic-alpine plants, in particular, could "run out of space" as they are out-competed by expansion of woody vegetation. Mountain regions could potentially provide safe sites for arctic-alpine plants in a warmer climate, but empirical evidence is fragmentary. Here we present a 24,000-year record of species persistence based on sedimentary ancient DNA (sedaDNA) from Lake Bolshoye Shchuchye (Polar Urals). We provide robust evidence of long-term persistence of arctic-alpine plants through large-magnitude climate changes but document a decline in their diversity during a past expansion of woody vegetation. Nevertheless, most of the plants that were present during the last glacial interval, including all of the arctic-alpines, are still found in the region today. This underlines the conservation significance of mountain landscapes via their provision of a range of habitats that confer resilience to climate change, particularly for arctic-alpine taxa.


Subject(s)
Climate Change , Ecosystem , Plant Development , Plants/classification , Arctic Regions
2.
J Mammary Gland Biol Neoplasia ; 24(3): 257-270, 2019 09.
Article in English | MEDLINE | ID: mdl-31104199

ABSTRACT

Germline mutations in the breast cancer susceptibility gene BRCA1, encoding a tumor suppressor protein, greatly enhance the risk of breast and ovarian cancer. This tissue-specificity implicates the role of ovarian hormones. Indeed, BRCA1 has been demonstrated to regulate the signalling axis of the hormone, progesterone, and its receptor, the progesterone receptor (PR), and progesterone action has been implicated in BRCA1-related tumorigenesis. BRCA1 also plays important roles in oxidative stress and activating nuclear factor kappaB (NFκB) signalling pathways. Like wildtype BRCA1 function, PR signalling has also been shown to inhibit NFκB activation. Although PR and BRCA1 networks are known to interact, their interaction at the level of NFκB activation in the human breast is not understood. This study investigates the effect of reduced BRCA1 expression on proliferation and NFκB activation in human breast cells, and the impact of progesterone on these effects. The major findings are that: 1) Reduced BRCA1 levels inhibit cell growth in normal human mammary cells and breast cancer cells; 2) Reduced BRCA1 levels stimulated inflammatory targets and NFκB activity in normal human mammary cells; 3) Wildtype BRCA1 inhibited the pro-proliferative effects of progesterone in normal mammary epithelial cells, and; 4) Progesterone attenuated BRCA1-mediated NFκB activation in normal human mammary cells. These data have important implications for our understanding of progesterone action in BRCA1 mutation carriers, and how inhibition of this action may potentially delay tumorigenesis or impart a more favourable prognosis.


Subject(s)
BRCA1 Protein/metabolism , Breast Neoplasms/pathology , Breast/pathology , Cell Proliferation , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , NF-kappa B/metabolism , Progesterone/pharmacology , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/genetics , Biomarkers, Tumor/genetics , Breast/drug effects , Breast/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic , Cells, Cultured , Female , Gene Expression Profiling , Humans , NF-kappa B/genetics , Progestins/pharmacology , RNA, Small Interfering/genetics , Signal Transduction
3.
Breast Cancer Res Treat ; 143(3): 423-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24395108

ABSTRACT

The epithelium of the human breast is made up of a branching ductal-lobular system, which is lined by a single layer of luminal cells surrounded by a contractile basal cell layer. The co-ordinated development of stem/progenitor cells into these luminal and basal cells is fundamentally important for breast morphogenesis. The ovarian steroid hormones, progesterone (P) and 17ß-estradiol, are critical in driving this normal breast development, yet ovarian activity has also been shown to be a major driver of breast cancer risk. We previously demonstrated that P treatment increases proliferation and augments the number of progenitor-like cells, and that the progesterone receptor (PR) is also expressed in the bipotent progenitor-enriched subfraction. Here we demonstrate that PR is expressed in a subset of CD10+ basal cells and that P stimulates this CD10+ cell compartment, which is enriched for bipotent progenitor activity. In addition, we have shown that P stimulates progenitor cells in human breast cancer cell lines and expands the cancer stem cell population via increasing the stem-like CD44+ population. As changes in cell type composition are one of the hallmark features of breast cancer progression, the demonstration that progenitor cells are stimulated by P in both normal breast and in breast cancer cells has critical implications in discerning the mechanisms of how P increases breast cancer risk.


Subject(s)
Breast Neoplasms/metabolism , Breast/drug effects , Cell Proliferation/drug effects , Progesterone/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Lineage , Estradiol/administration & dosage , Estradiol/genetics , Female , Humans , Hyaluronan Receptors/genetics , Neoplastic Stem Cells/drug effects , Neprilysin/genetics , Progesterone/genetics , Receptors, Estrogen/metabolism , Stem Cells/drug effects
4.
Eur J Vasc Endovasc Surg ; 45(2): 162-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23265683

ABSTRACT

OBJECTIVE: To develop a method of event-based analysis that quantifies the fragmented nature of walking bouts in individuals with intermittent claudication [IC] and compare outcomes with age and gender-matched healthy controls. DESIGN: Cross-sectional. MATERIALS: The activPAL™ physical activity monitor. METHODS: 7-day physical activity patterns were compared between individuals with IC (n = 30) and controls matched for age and gender (n = 30). The ratio of the number of walking events to upright events was calculated to provide an event-based claudication index (EBCI) that represented the fragmented nature of walking bouts commonly reported in those with IC. RESULTS: Individuals with IC had a greater EBCI than age matched controls indicating a more fragmented walking pattern (5.8 ± 2.0 vs. 7.7 ± 3.1, p < 0.01). The difference between groups was more pronounced when the EBCI was calculated from upright events that included >400 steps (23.4 ± 11.3 vs. 35.8 ± 14.2, p < 0.01). CONCLUSION: The classic fragmented stop/start walking pattern universally described by individuals with IC can be quantified using the EBCI. This method of measurement potentially provides a novel method of assessing the effectiveness of clinical interventions for this patient group.


Subject(s)
Actigraphy , Intermittent Claudication/diagnosis , Motor Activity , Walking , Actigraphy/instrumentation , Aged , Ankle Brachial Index , Case-Control Studies , Cross-Sectional Studies , Female , Humans , Intermittent Claudication/physiopathology , Male , Middle Aged , Predictive Value of Tests , Prognosis , Severity of Illness Index , Time Factors
5.
Mol Cell Endocrinol ; 361(1-2): 191-201, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22580007

ABSTRACT

Progesterone is critical in normal breast development and its synthetic derivatives are emerging as major drivers of breast cancer risk. The recent demonstration that progesterone regulates the stem cell compartment in the murine mammary gland, despite the absence of progesterone receptor (PR) in mammary stem cells, highlights the fact that PR distribution in progenitor cell subsets in the human breast remains to be conclusively shown. By utilising two independent cell sorting strategies to fractionate cells into distinct subpopulations enriched for different cell lineage characteristics, we have demonstrated a consistent enrichment of PR transcripts, relative to estrogen receptor transcripts, in the bipotent progenitor subfraction in the normal human breast. We have also shown co-expression of both steroid hormone receptors with basal markers in a subset of human breast cells, and finally we have demonstrated that PR+ bipotent progenitor cells are estrogen-insensitive, and that estrogen regulates PR in mature luminal cells only.


Subject(s)
Breast/cytology , Breast/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Adult , Animals , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation/drug effects , Cell Separation , Cells, Cultured , Epithelial Cell Adhesion Molecule , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estrogens/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Integrin alpha6/metabolism , Keratin-14/metabolism , Mice , Middle Aged , Models, Biological , NIH 3T3 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Young Adult
6.
Mol Endocrinol ; 24(7): 1380-92, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20519331

ABSTRACT

Prolactin and progesterone act together to regulate mammary alveolar development, and both hormones have been implicated in breast cancer initiation and progression. Here we show that Elf5, a prolactin-induced ETS transcription factor that specifies the mammary secretory cell lineage, is also induced by progestins in breast cancer cells via a direct mechanism. To define the transcriptional response to progestin elicited via Elf5, we made an inducible Elf5 short hairpin-RNA knock-down model in T47D breast cancer cells and used it to prevent the progestin-induction of Elf5. Functional analysis of Affymetrix gene expression data using Gene Ontologies and Gene Set Enrichment Analysis showed enhancement of the progestin effects on cell cycle gene expression. Cell proliferation assays showed a more efficacious progestin-induced growth arrest when Elf5 was kept at baseline levels. These results showed that progestin induction of Elf5 expression tempered the antiproliferative effects of progestins in T47D cells, providing a further mechanistic link between prolactin and progestin in the regulation of mammary cell phenotype.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Progestins/pharmacology , Progestins/therapeutic use , Proto-Oncogene Proteins c-ets/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA-Binding Proteins , Female , Humans , Mifepristone/pharmacology , Oligonucleotide Array Sequence Analysis , RNA Interference , Transcription Factors
7.
Clin Pharmacol Ther ; 82(1): 33-40, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17495881

ABSTRACT

The aim of this study was to explore the impact of individual variation in drug elimination on imatinib disposition. Twenty-two patients with gastrointestinal stromal tumor or chronic myeloid leukemia initially received imatinib 600 mg daily with dosage subsequently toxicity adjusted. Pharmacokinetic parameters on day 1 and at steady-state were compared with elimination phenotype and single-nucleotide polymorphisms of CYP3A5 and ABCB1. A fivefold variation in estimated imatinib clearance (CL/F) was present on day 1 and mean CL/F had fallen by 26% at steady state. This reduction in imatinib CL/F was associated with ABCB1 genotype, being least apparent in thymidine homozygotes at the 1236T>C, 2677G>T/A and 3435C>T loci. Toxicity-related dose reduction also tended to be less common in these individuals. ABCB1 genotype was associated with steady-state CL/F due to an apparent genotype-specific influence of imatinib on elimination. Further evaluation of ABCB1 genotype and imatinib dosage is warranted.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Gastrointestinal Stromal Tumors/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Organic Anion Transporters/genetics , Piperazines/pharmacokinetics , Polymorphism, Single Nucleotide , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Benzamides , Cohort Studies , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Drug Monitoring , Female , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Genotype , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Male , Metabolic Clearance Rate , Middle Aged , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/metabolism , Phenotype , Piperazines/administration & dosage , Piperazines/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects
8.
Article in English | MEDLINE | ID: mdl-18540569

ABSTRACT

Progesterone is an essential regulator of normal female reproductive function, yet recent studies on the use of progestins in hormone replacement therapy have clearly implicated progestins in breast cancer development, a disease initiated early in life at a time of frequent exposure to cycling ovarian hormones. The effects of progesterone are mediated by two distinct nuclear receptor proteins, PRA and PRB. In normal breast PRA and PRB are co-expressed at similar levels in luminal epithelial cells, suggesting that both proteins are required to mediate physiologically relevant progesterone signalling. However, early in breast carcinogenesis PRA:PRB expression is disrupted, resulting in frequent predominance of one isoform. Unbalanced expression of PRA and PRB results in altered hormonal response and aberrant targeting of genes that are not normally progestin-regulated, principally those involved in morphological changes and disruptions of the actin cytoskeleton, and in migration. Movement of PR into discrete nuclear domains, or foci, is a critical step in normal PR transcriptional activity that appears to be aberrant in cancers and likely related to alterations in nuclear morphology, gene expression and cell function associated with tumour cells. Given that exogenous progestins are consumed by millions of women worldwide in the form of hormone replacement therapy and oral contraceptives, it is vital to better understand the mechanisms of progesterone action in the breast.


Subject(s)
Breast Neoplasms/physiopathology , Breast/physiology , Protein Isoforms/physiology , Receptors, Progesterone/physiology , Animals , Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Humans , Ligands , Protein Isoforms/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Transcription, Genetic/physiology
9.
Br J Cancer ; 95(4): 515-9, 2006 Aug 21.
Article in English | MEDLINE | ID: mdl-16880780

ABSTRACT

We have studied loss of heterozygosity at the BRCA1 and BRCA2 loci in 992 normal cell clones derived from topographically defined areas of normal tissue in four samples from BRCA1/BRCA2 mutation carriers. The frequency of loss of heterozygosity in the clones was low (1.01%), but it was found in all four samples, whether or not a tumour was present. Topographical mapping revealed that the genetic changes were clustered in some breast samples. Our study confirms the previous finding that a field of genetic instability can exist around a tumour, suggesting that sufficient tissue must be removed at surgery to avoid local recurrence. We also demonstrate that such a field of genetic change can exist in morphologically normal tissue before a tumour develops and, for the first time, we demonstrate that the field is of a size greater than one terminal duct-lobular unit. The genetic changes are not identical, however, which suggests that genetic instability in these regions may play an early role in tumour development. We also confirm and extend our original observation of loss of the wild-type BRCA1 allele in some clones, and loss of the mutant allele in others, demonstrating that loss of either allele is a stochastic event.


Subject(s)
Breast/cytology , Genes, BRCA1 , Genes, BRCA2 , Genetic Predisposition to Disease , Breast Neoplasms/genetics , Clone Cells , Female , Gene Frequency , Humans , Loss of Heterozygosity , Mastectomy , Mutation
10.
Oncogene ; 25(58): 7597-607, 2006 Dec 07.
Article in English | MEDLINE | ID: mdl-16799644

ABSTRACT

Reports that the adhesion-associated molecule p130Cas/BCAR1 promotes resistance to tamoxifen suggested that adhesion-mediated signalling may be altered by tamoxifen treatment. We find that p130Cas/BCAR1 phosphorylation is enhanced in tamoxifen-treated estrogen receptor (ER)-positive MCF-7 breast cancer cells. The effects of estrogen and tamoxifen were assessed independently and in combination, and the results demonstrate that tamoxifen antagonizes estrogen regulation of p130Cas/BCAR1 phosphorylation. Phosphorylation correlates with tamoxifen ER antagonist effects, as phosphorylation effects are replicated by the pure antiestrogen ICI 182, 780. Correspondingly, phosphorylation is not changed in ER-negative cells exposed to tamoxifen. We show that deletion of the p130Cas/BCAR1 substrate domain substantially reduces tamoxifen-induced phosphorylation of p130Cas/BCAR1 and confers enhanced sensitivity to tamoxifen. P130Cas/BCAR1 forms a phosphorylation-dependent signalling complex with focal adhesion kinase (FAK) and Src kinase that promotes adhesion-mediated cell survival. Therefore, we examined the kinetics of p130Cas/BCAR1, Src and FAK phosphorylation over a 14-day time course and find sustained phosphorylation of these molecules after 7 days exposure to tamoxifen. Inhibition of Src kinase is shown to reduce tamoxifen-promoted p130Cas/BCAR1 phosphorylation and reduce cell viability. Stimulation of the Src/FAK/p130Cas/BCAR1 adhesion signalling pathway in tamoxifen-treated MCF-7 cells does not cause increased migration; however, there is Src-dependent phosphorylation of the cell survival molecule Akt. Correspondingly, Akt inhibition reduces cell viability in cells treated with tamoxifen. We propose that prolonged activation of adhesion-dependent signalling may confer a survival advantage in response to additional cellular insults or alternatively, may poise cells to develop a migratory phenotype in response to additional cellular cues.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cell Adhesion Molecules/metabolism , Crk-Associated Substrate Protein/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Tamoxifen/pharmacology , src-Family Kinases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptors, Estrogen/metabolism
11.
Br J Cancer ; 92(8): 1366-71, 2005 Apr 25.
Article in English | MEDLINE | ID: mdl-15812557

ABSTRACT

Histopathologic features of breast cancer such as tumour size, grade and axillary lymph node (LN) status variably reflect tumour biology and time. Recent evidence suggests that the biological character of breast cancer is established at an early stage and has a major impact on clinical course. The aim of this study was to distinguish the impact of biology on breast cancer histopathology by comparing features of breast cancers diagnosed following population mammographic screening with prevalent vs incident detection and screening interval. Central histopathology review data from 1147 cases of ductal in situ and/or invasive breast cancer were examined. Size, grade and LN status of invasive cancers were positively correlated (P < 0.001). Prevalent invasive cancers were larger (P < 0.001) and more likely to be LN positive (P = 0.02) than incident cases, but grade was not associated with screening episode (P = 0.7). Screening interval for incident cancers was positively associated with invasive cancer size (P = 0.05) and LN status (P = 0.002) but not grade (P = 0.1). Together, these data indicate that biology and time both impact on size and LN status of invasive breast cancer, but grade reflects biology alone. In view of the clinical importance of breast cancer biology, grade as its most direct indicator assumes particular significance.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Mass Screening , Adult , Aged , Aged, 80 and over , Female , Humans , Lymphatic Metastasis/pathology , Mammography , Middle Aged , Time Factors
12.
Breast Cancer Res Treat ; 83(3): 211-20, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14758091

ABSTRACT

Women exposed to exogenous progesterone have increased breast cancer risk, but the mechanisms of progesterone involvement in breast cancer development are unknown. In human breast and endometrium, progesterone receptor (PR) isoform expression is disrupted in premalignant lesions and predominance of one isoform, usually PRA, in invasive cancers is associated with poorer prognosis. Disrupted PR isoform expression results in disrupted progestin regulation of cell morphology, including rounded morphology and decreased adherence of cells to tissue culture flasks. The purpose of this study was to test the hypothesis that predominance of PRA affects the interaction of breast cancer cells with a physiologically relevant stromal tissue, bone marrow stroma. T-47D breast cancer cells demonstrated the ability to migrate into bone marrow fibroblasts and this was inhibited by progestin treatment. The antiprogestin RU38486 abrogated the progestin effect on migration, demonstrating that it was PR-mediated. In cells expressing a predominance of PRA, after induction of a stably integrated inducible PRA construct, the ability of progestin to inhibit breast cancer cell migration was lost. A number of integrins were progestin regulated in T-47D cells, but there was no difference in the progestin effect in cells with PRA predominance, nor were the levels of focal adhesion proteins altered in these cells. This suggested that the lack of inhibition by progestin of breast cancer cell migration in cells with PRA predominance was not mediated by PRA effects on the membrane components of the adherens junctions. In summary, this study has shown that PRA predominance has a striking functional effect on breast cancer cell migration into stromal layers. PRA predominance may render breast cancer cells relatively resistant to the inhibitory effects of progestins and one consequence of this may be increased invasion of stroma. If borne out in vivo, these findings suggest that tumours with PRA predominance may be predisposed to cancer progression and this may signal a poorer prognosis in patients.


Subject(s)
Bone Marrow Neoplasms/secondary , Breast Neoplasms/pathology , Cell Movement , Neoplasm Metastasis/physiopathology , Receptors, Progesterone/analysis , Female , Fibroblasts , Humans , Integrins/physiology , Neoplastic Cells, Circulating , Progestins/pharmacology , Prognosis , Risk Factors , Tumor Cells, Cultured
13.
J Mol Endocrinol ; 31(2): 241-53, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14519093

ABSTRACT

Changes in the cell cytoskeleton occur in cell transformation and recent data suggest the involvement of ovarian hormones, which are implicated in cancer development and progression. In human breast and endometrial tumors, there is disrupted expression of progesterone receptor (PR) isoforms and predominance of one isoform, usually PRA. PRA predominance is an early event in carcinogenesis, and in cancers is associated with poor clinical features. Overexpression of PRA in vitro causes altered progestin regulation of cell morphology, suggesting that PRA overexpression may provoke deleterious changes in cell functioning. This study aimed to identify pathways of cytoskeleton regulation responsive to progestins and to determine whether these are perturbed when PRA is overexpressed to the levels seen in cancers. Progestin treatment of PR-positive breast cancer cells caused increased cell surface area whereas after induction of a stably integrated PRA construct, cells became rounded and the cell surface was decreased. The effect of PRA induction on cell rounding was reversed by the anti-progestin RU38486. Altered tropomyosin (Tm) isoforms were implicated in these morphological differences, as there was a PRA-mediated alteration in Tm5 isoform levels, and transfection of Tm5a mimicked progestin-mediated cell rounding in PRA-overexpressing cells. Ezrin was redistributed from the membrane to cytoplasmic locations in the presence of progestin, and discrete focal localization was evident in cells with PRA predominance. Progestin effects on the cytoskeleton in PRA-overexpressing cells provide evidence for novel endocrine regulation of aspects of actin microfilament composition, suggesting that changes in the cytoskeleton known to be associated with cancer development and progression may be regulated in part by altered PRA expression which develops early in carcinogenesis.


Subject(s)
Actin Cytoskeleton/metabolism , Progestins/metabolism , Receptors, Progesterone/metabolism , Signal Transduction/physiology , Actins/metabolism , Animals , Cytoskeletal Proteins/metabolism , Female , Focal Adhesions/metabolism , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Phosphoproteins/metabolism , Progestins/pharmacology , Rats , Signal Transduction/drug effects , Trans-Activators/metabolism , beta Catenin
14.
Breast Cancer Res Treat ; 72(2): 163-72, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12038707

ABSTRACT

Progesterone receptor (PR) mediates the effects of progesterone in mammary tissues and plays a crucial role in normal breast development and in breast cancer. PR proteins are expressed as two isoforms, PRA and PRB, that have different capacities to activate target genes, yet it is unknown whether progesterone action in normal and malignant breast is mediated by PRA and/or PRB. This study determines the relative expression of PRA and PRB in normal breast and in benign, premalignant and malignant archival breast lesions by dual immunofluorescent histochemistry. In normal breast and in proliferative disease without atypia (PDWA) PRA and PRB were co-expressed within the same cells in comparable amounts, implicating both isoforms in progesterone action. In atypical lesions, however, there was a significant increase in predominant expression of PRA or PRB, with lesion progression from the normal state to malignancy. PR isoform predominance, especially PRA predominance, was evident in a high proportion of ductal carcinomas in situ (DCIS) and invasive breast lesions. In the normal breast and in PDWA, the relative expression of PRA and PRB in adjacent cells was homogenous. There was a significant increase in cell-to-cell heterogeneity of PR isoform expression in ADH and DCIS lesions and in the majority of breast cancers. Heterogeneous cell-to-cell expression of PR isoforms occurred prior to overall predominant expression of one isoform in premalignant breast lesions, demonstrating that loss of control of relative PRA:PRB expression is an early event in the development of breast cancer. PRA:PRB ratios within a breast lesion are likely to be important as both markers and effectors of tumor growth and development, and progressively aberrant PR isoform expression may play a role in the etiology of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Receptors, Progesterone/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Breast Diseases/metabolism , Female , Humans , Middle Aged , Time Factors
17.
J Clin Pathol ; 54(8): 624-30, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11477119

ABSTRACT

AIM: The measurement of progesterone receptors (PR) is recommended as part of the clinical management of breast and endometrial cancers, and immunohistochemistry on formalin fixed tissue is now the method of choice. PR is expressed as two isoforms, PRA and PRB, and although both these proteins are expressed in hormone dependent cancers, there is evidence that a large proportion of tumours express a predominance of one isoform. Therefore, it is essential to document the individual detection of PRA and PRB by the presently available anti-PR antibodies. The aim of this study is to investigate the detection of PR isoforms A and B in formalin fixed, paraffin wax embedded cell lines and tissue sections by immunohistochemistry, using a panel of commercial and in house antibodies to human PR. METHODS: PR negative cell lines stably transfected to express only PRA (MCF-7Mll/PRA) or PRB (MDA-MB-231/PRB), and tissue sections of human breast carcinoma and normal endometrium were stained using an immunoperoxidase method. A panel of primary PR specific antibodies was evaluated for ability to detect both PRA and PRB proteins, and for intensity and distribution of positive staining under optimal conditions. RESULTS: Of the 11 antibodies assessed, only four recognised PRA and PRB similarly. Six recognised PRA proteins but were unable to detect PRB expression in the cell lines expressing only PRA or PRB. In tissues expressing high amounts of PRA and PRB, all antibodies tested demonstrated positive PR staining. However, in tissues expressing a predominance of PRB, differential staining patterns were observed, with variations in staining intensity and in the proportion of cells positive for PR. CONCLUSIONS: Most PR specific antibodies tested failed to detect PRB in formalin fixed tissue by immunohistochemical techniques, despite their ability to do so by immunoblot analysis. These observations suggest that there are conformational differences between PRA and PRB that mask epitopes on the PRB protein recognised by most anti-PR antibodies. The selection of antibodies that recognise both PRB and PRA in formalin fixed tissue is essential for the accurate evaluation of PR positivity in clinical specimens.


Subject(s)
Receptors, Progesterone/analysis , Animals , Antibodies , Breast Neoplasms/chemistry , COS Cells , Carcinoma, Ductal, Breast/chemistry , Endometrium/chemistry , Female , Formaldehyde , Humans , Immunoblotting , Immunoenzyme Techniques , Rabbits , Receptors, Progesterone/genetics , Sensitivity and Specificity , Tissue Fixation , Transfection , Tumor Cells, Cultured/chemistry
18.
J Adv Nurs ; 34(2): 264-72, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11430289

ABSTRACT

BACKGROUND: Organizational and professional learning are interrelated processes that underpin the contemporary drive for a quality evidence-based delivery of health care in the United Kingdom (UK). DESIGN: A soft systems methodology was used to explore the pervasiveness of practice developments. Three case study sites were identified using matrix sampling and data collected through 29 individual interviews and two focus group interviews, with the interviews augmented with a tool designed to maximize analysis of the processes of developing practice. FINDINGS: The resultant model of developing health care practice includes three processes: using and creating knowledge, understanding and practice of patient care, and effecting development. The whole model was underpinned by professional and organizational learning in which 'expert thinkers' engaged in double loop learning to reconceptualize care rather than just perpetuate existing patterns of care delivery.


Subject(s)
Attitude of Health Personnel , Education, Nursing, Continuing/organization & administration , Health Knowledge, Attitudes, Practice , Models, Nursing , Nurses/psychology , Nursing Evaluation Research/organization & administration , Professional Autonomy , Research Personnel/psychology , Staff Development/organization & administration , Evidence-Based Medicine , Focus Groups , Health Services Research , Humans , Learning , Models, Educational , Models, Organizational , Organizational Culture , Surveys and Questionnaires , Systems Analysis , United Kingdom
19.
Cancer Res ; 61(11): 4576-82, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389093

ABSTRACT

The nuclear receptor for the female hormone progesterone (PR) is widely expressed in uterine cancer. PR is expressed as two proteins (PRA and PRB) with different functions, and in vitro evidence reveals PRA to inhibit PRB function, so the cellular ratio of PRA:PRB is likely to be an important determinant of progesterone action. The relative expression of PRA and B and their involvement in the pathogenesis of endometrial cancer is not known. The aims of this study were to determine PRA and B expression by dual immunofluorescent histochemistry in endometrial adenocarcinomas compared with expression in normal and hyperplastic glands, and to correlate expression in tumors with clinical features including grade. Significantly lower PR levels were found in tumors compared with normal glands and areas of complex atypical hyperplasia within the same specimen. The normal glands expressed both of the isoforms at similar levels, whereas there was increased predominance of one isoform in hyperplastic areas and in tumors, which suggested that the loss of coordinated expression of PR isoforms was an early event in tumor progression. The majority of tumors [27 (58%) of 46] expressed only one PR isoform, and the proportion expressing either PRA or B was the same [14 (30%) of 46, and 13 (28%) of 46, respectively]. One-half of all tumors ([23 (50%) of 46] expressed either PRA only or a predominance of PRA, and a few tumors [10 (22%) of 46] expressed comparable levels of PRA and B. Similar levels of PRA and B were noted only in FIGO grade 1 tumors, whereas higher grades (2 and 3) were associated with a predominance of one isoform. In summary, expression of only one PR isoform was common in endometrial cancers, which indicates that the decreased PR levels observed in these cancers arise from the loss of one PR isoform. Expression of a single PR isoform was associated with higher clinical grade, which suggests a relationship between the loss of PR isoform expression and features of poorer prognosis. Disruption of relative PR isoform expression was observed in complex atypical hyperplasia, which suggests that early alterations in the ratio of PRA:PRB may precede and/or be implicated in the development of endometrial adenocarcinoma. Alterations in the ratio of PR isoform expression are likely to cause disordered regulation of target genes, resulting in altered progestin action in the uterus, and this may be involved in the pathogenesis of endometrial cancer.


Subject(s)
Carcinoma, Endometrioid/metabolism , Endometrial Neoplasms/metabolism , Receptors, Progesterone/biosynthesis , Adult , Aged , Aged, 80 and over , Carcinoma, Endometrioid/pathology , Cohort Studies , Endometrial Hyperplasia/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Middle Aged , Neoplasm Staging , Protein Isoforms
20.
Clin Cancer Res ; 6(11): 4334-42, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11106251

ABSTRACT

Full-term pregnancy early in reproductive life is protective against breast cancer in women. The protective effects of parity have variously been attributed to the differentiation that accompanies pregnancy and lactation, alterations in ovarian hormone receptor levels, and altered sensitivity to ovarian hormones. Butyrate, a short-chain fatty acid, induces differentiation in breast cancer cell lines and decreases hormone receptor expression. Butyrate also inhibits proliferation in breast cancer cell lines and modulates expression of key cell cycle-regulatory proteins including cyclin D1. Given these properties, butyrate could be considered a promising agent for breast cancer prevention. Therefore, this study aimed to determine the effects of butyrate on normal human breast epithelial cells and to compare the effects of two stable butyrate derivatives with more favorable pharmacological properties: phenylacetate and its p.o. active precursor phenylbutyrate. Treatment with each agent resulted in concentration-dependent growth inhibition in a normal breast epithelial cell line and two breast cancer cell lines (MCF-7 and MDA-MB-231). Phenylbutyrate and butyrate inhibited proliferation to a similar extent, but phenylacetate was less effective in all of the cell lines. All three of the agents induced differentiation (accumulation of lipid droplets) in normal as well as in breast cancer cells and caused a decrease in estrogen receptor (ER) mRNA in MCF-7 cells. The butyrates decreased expression of cyclin D1, increased expression of p21(Waf1/Cip1), and hypophosphorylated pRB in the normal mammary epithelial cells. The effects on cyclin D1 expression correlated with the effects on cell proliferation, which suggests that modulation of cyclin D1 expression may underpin the antiproliferative effects of butyrates. We have shown that butyrate and butyrate-like agents are able to decrease proliferation and induce differentiation in normal breast cells as well as in malignant breast cells (ER-positive and ER-negative) and, as such, may be considered as candidate chemopreventative agents for women at high risk of developing breast cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast/drug effects , Butyrates/pharmacology , Cyclin D1/genetics , Cyclins/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors , Breast/cytology , Breast/metabolism , Cell Differentiation/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Genes, Retinoblastoma , Humans , RNA, Messenger/analysis , Receptors, Estrogen/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...